Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Biol Macromol ; 280(Pt 1): 135700, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39288862

RESUMEN

SARS-CoV-2 pandemic clearly demonstrated the lack of preparation against novel and emerging viral diseases. This prompted an enormous effort to identify antivirals to curb viral spread and counteract future pandemics. Ribosome Inactivating Proteins (RIPs) and Ribotoxin-Like Proteins (RL-Ps) are toxin enzymes isolated from edible plants and mushrooms, both able to inactivate protein biosynthesis. In the present study, we combined imaging analyses, transcriptomic and proteomic profiling to deeper investigate the spectrum of antiviral activity of quinoin, type 1 RIP from quinoa seeds. Here, we show that RIPs, but not RL-Ps, act on a post-entry step and impair SARS-CoV-2 replication, potentially by direct degradation of viral RNA. Interestingly, the inhibitory activity of quinoin was conserved also against other members of the Coronaviridae family suggesting a broader antiviral effect. The integration of mass spectrometry (MS)-based proteomics with transcriptomics, provided a comprehensive picture of the quinoin dependent remodeling of crucial biological processes, highlighting an unexpected impact on lipid metabolism. Thus, direct and indirect mechanisms can contribute to the inhibitory mechanism of quinoin, making RIPs family a promising candidate not only for their antiviral activity, but also as an effective tool to better understand the cellular functions and factors required during SARS-CoV-2 replication.

2.
Neurobiol Dis ; 200: 106622, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39097034

RESUMEN

The complexity and heterogeneity of PD necessitate advanced diagnostic and prognostic tools to elucidate its molecular mechanisms accurately. In this study, we addressed this challenge by conducting a pilot phospho-proteomic analysis of peripheral blood mononuclear cells (PBMCs) from idiopathic PD patients at varying disease stages to delineate the functional alterations occurring in these cells throughout the disease course and identify key molecules and pathways contributing to PD progression. By integrating clinical data with phospho-proteomic profiles across various PD stages, we identify potential stage-specific molecular signatures indicative of disease progression. This integrative approach allows for the discernment of distinct disease states and enhances our understanding of PD heterogeneity.


Asunto(s)
Progresión de la Enfermedad , Leucocitos Mononucleares , Enfermedad de Parkinson , Proteoma , Proteómica , Humanos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/patología , Leucocitos Mononucleares/metabolismo , Proteoma/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Proteómica/métodos , Anciano , Fosfoproteínas/metabolismo
3.
NPJ Syst Biol Appl ; 10(1): 95, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39179556

RESUMEN

Unraveling how cellular signaling is remodeled upon perturbation is crucial for understanding disease mechanisms and identifying potential drug targets. In this pursuit, computational tools generating mechanistic hypotheses from multi-omics data have invaluable potential. Here, we present a newly implemented version (2.0) of SignalingProfiler, a multi-step pipeline to draw mechanistic hypotheses on the signaling events impacting cellular phenotypes. SignalingProfiler 2.0 derives context-specific signaling networks by integrating proteogenomic data with the prior knowledge-causal network. This is a freely accessible and flexible tool that incorporates statistical, footprint-based, and graph algorithms to accelerate the integration and interpretation of multi-omics data. Through a benchmarking process on three proof-of-concept studies, we demonstrate the tool's ability to generate hierarchical mechanistic networks recapitulating novel and known perturbed signaling and phenotypic outcomes, in both human and mice contexts. In summary, SignalingProfiler 2.0 addresses the emergent need to derive biologically relevant information from complex multi-omics data by extracting interpretable networks.


Asunto(s)
Algoritmos , Biología Computacional , Fenotipo , Transducción de Señal , Humanos , Transducción de Señal/genética , Transducción de Señal/fisiología , Ratones , Animales , Biología Computacional/métodos , Programas Informáticos , Proteogenómica/métodos , Multiómica
4.
Elife ; 122024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38564252

RESUMEN

Currently, the identification of patient-specific therapies in cancer is mainly informed by personalized genomic analysis. In the setting of acute myeloid leukemia (AML), patient-drug treatment matching fails in a subset of patients harboring atypical internal tandem duplications (ITDs) in the tyrosine kinase domain of the FLT3 gene. To address this unmet medical need, here we develop a systems-based strategy that integrates multiparametric analysis of crucial signaling pathways, and patient-specific genomic and transcriptomic data with a prior knowledge signaling network using a Boolean-based formalism. By this approach, we derive personalized predictive models describing the signaling landscape of AML FLT3-ITD positive cell lines and patients. These models enable us to derive mechanistic insight into drug resistance mechanisms and suggest novel opportunities for combinatorial treatments. Interestingly, our analysis reveals that the JNK kinase pathway plays a crucial role in the tyrosine kinase inhibitor response of FLT3-ITD cells through cell cycle regulation. Finally, our work shows that patient-specific logic models have the potential to inform precision medicine approaches.


Asunto(s)
Leucemia Mieloide Aguda , Transducción de Señal , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Sistema de Señalización de MAP Quinasas , Línea Celular , Resistencia a Medicamentos , Tirosina Quinasa 3 Similar a fms/genética
5.
Br J Cancer ; 129(11): 1707-1716, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37898722

RESUMEN

The Cyclin-dependent kinase 1, as a serine/threonine protein kinase, is more than a cell cycle regulator as it was originally identified. During the last decade, it has been shown to carry out versatile functions during the last decade. From cell cycle control to gene expression regulation and apoptosis, CDK1 is intimately involved in many cellular events that are vital for cell survival. Here, we provide a comprehensive catalogue of the CDK1 upstream regulators and substrates, describing how this kinase is implicated in the control of key 'cell cycle-unrelated' biological processes. Finally, we describe how deregulation of CDK1 expression and activation has been closely associated with cancer progression and drug resistance.


Asunto(s)
Proteína Quinasa CDC2 , Proteínas Serina-Treonina Quinasas , Humanos , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Genes cdc , Ciclo Celular , División Celular
6.
Anal Chem ; 95(24): 9199-9206, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37278511

RESUMEN

The assessment of the higher-order structure (HOS) by NMR is a powerful methodology to characterize the structural features of biologics. Forced oxidative stress studies are used to investigate the stability profile, to develop pharmaceutical formulations and analytical methods. Here, the effects of forced oxidative stress by H2O2 on the monoclonal antibody Abituzumab have been characterized by a multianalytical approach combining NMR spectroscopy, mass spectrometry, differential scanning calorimetry, surface plasmon resonance, computational tools, and bioassays. This integrated strategy has provided qualitative and semiquantitative characterization of the samples and information at residue level of the effects that oxidation has on the HOS of Abituzumab, correlating them to the loss of the biological activity.


Asunto(s)
Anticuerpos Monoclonales , Peróxido de Hidrógeno , Flujo de Trabajo , Anticuerpos Monoclonales/química , Espectroscopía de Resonancia Magnética
8.
Cell Death Differ ; 30(2): 417-428, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36460775

RESUMEN

Caspase-8 is a cysteine protease that plays an essential role in apoptosis. Consistently with its canonical proapoptotic function, cancer cells may genetically or epigenetically downregulate its expression. Unexpectedly, Caspase-8 is often retained in cancer, suggesting the presence of alternative mechanisms that may be exploited by cancer cells to their own benefit. In this regard, we reported that Src tyrosine kinase, which is aberrantly activated in many tumors, promotes Caspase-8 phosphorylation on Tyrosine 380 (Y380) preventing its full activation. Here, we investigated the significance of Caspase-8 expression and of its phosphorylation on Y380 in glioblastoma, a brain tumor where both Caspase-8 expression and Src activity are often aberrantly upregulated. Transcriptomic analyses identified inflammatory response as a major target of Caspase-8, and in particular, NFκB signaling as one of the most affected pathways. More importantly, we could show that Src-dependent phosphorylation of Caspase-8 on Y380 drives the assembly of a multiprotein complex that triggers NFκB activation, thereby inducing the expression of inflammatory and pro-angiogenic factors. Remarkably, phosphorylation on Y380 sustains neoangiogenesis and resistance to radiotherapy. In summary, our work identifies a novel interplay between Src kinase and Caspase-8 that allows cancer cells to hijack Caspase-8 to sustain tumor growth.


Asunto(s)
Caspasa 8 , Glioblastoma , Familia-src Quinasas , Humanos , Apoptosis , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Glioblastoma/genética , Fosforilación , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo
9.
Leukemia ; 37(2): 288-297, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36509894

RESUMEN

The insertion site of the internal tandem duplications (ITDs) in the FLT3 gene affects the sensitivity to tyrosine kinase inhibitors (TKIs) therapy in acute myeloid leukemia (AML). Patients with the ITD in the tyrosine kinase domain lack effective therapeutic options. Here, to identify genotype-driven strategies increasing the TKI therapy efficacy, we developed SignalingProfiler, a strategy supporting the integration of high-sensitive mass spectrometry-based (phospho)proteomics, RNA sequencing datasets with literature-derived signaling networks. The approach generated FLT3-ITD genotype-specific predictive models and revealed a conserved role of the WEE1-CDK1 axis in TKIs resistance. Remarkably, pharmacological inhibition of the WEE1 kinase synergizes and strengthens the pro-apoptotic effect of TKIs therapy in cell lines and patient-derived primary blasts. Finally, we propose a new molecular mechanism of TKIs resistance in AML and suggest the combination of WEE1 inhibitor and TKI as a therapeutic option to improve patients clinical outcome.


Asunto(s)
Leucemia Mieloide Aguda , Inhibidores de Proteínas Quinasas , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Resistencia a Antineoplásicos/genética , Línea Celular , Transducción de Señal , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Mutación , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Proteína Quinasa CDC2/farmacología
10.
Rev Med Suisse ; 18(807): 2352, 2022 12 07.
Artículo en Francés | MEDLINE | ID: mdl-36477287

Asunto(s)
Médicos , Humanos
11.
Sci Adv ; 8(35): eabo1215, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36044577

RESUMEN

Selective degradation of the endoplasmic reticulum (ER) via autophagy (ER-phagy) is initiated by ER-phagy receptors, which facilitate the incorporation of ER fragments into autophagosomes. FAM134 reticulon family proteins (FAM134A, FAM134B, and FAM134C) are ER-phagy receptors with structural similarities and nonredundant functions. Whether they respond differentially to the stimulation of ER-phagy is unknown. Here, we describe an activation mechanism unique to FAM134C during starvation. In fed conditions, FAM134C is phosphorylated by casein kinase 2 (CK2) at critical residues flanking the LIR domain. Phosphorylation of these residues negatively affects binding affinity to the autophagy proteins LC3. During starvation, mTORC1 inhibition limits FAM134C phosphorylation by CK2, hence promoting receptor activation and ER-phagy. Using a novel tool to study ER-phagy in vivo and FAM134C knockout mice, we demonstrated the physiological relevance of FAM134C phosphorylation during starvation-induced ER-phagy in liver lipid metabolism. These data provide a mechanistic insight into ER-phagy regulation and an example of autophagy selectivity during starvation.

12.
Chem Sci ; 13(20): 5860-5871, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35685802

RESUMEN

A common criterion for designing performant single molecule magnets and pseudocontact shift tags is a large magnetic anisotropy. In this article we present a dysprosium complex chemically designed to exhibit strong easy-axis type magnetic anisotropy that is preserved in dichloromethane solution at room temperature. Our detailed theoretical and experimental studies on the magnetic properties allowed explaining several features typical of highly performant SMMs. Moreover, the NMR characterization shows remarkably large chemical shifts, outperforming the current state-of-the art PCS tags.

13.
Methods Mol Biol ; 2456: 123-140, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35612739

RESUMEN

Over the recent years, mass spectrometry (MS)-based proteomics has undergone dramatic advances in sample preparation, instrumentation, and computational methods. Here, we describe in detail, how a workflow quantifies global protein phosphorylation in pancreatic islets and characterizes intracellular organelle composition on protein level by MS-based proteomics.


Asunto(s)
Islotes Pancreáticos , Proteómica , Islotes Pancreáticos/metabolismo , Espectrometría de Masas/métodos , Orgánulos/metabolismo , Fosforilación , Proteómica/métodos
14.
J Am Chem Soc ; 144(22): 10006-10016, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35617699

RESUMEN

Multispecific biologics are an emerging class of drugs, in which antibodies and/or proteins designed to bind pharmacological targets are covalently linked or expressed as fusion proteins to increase both therapeutic efficacy and safety. Epitope mapping on the target proteins provides key information to improve the affinity and also to monitor the manufacturing process and drug stability. Solid-state NMR has been here used to identify the pattern of the residues of the programmed cell death ligand 1 (PD-L1) ectodomain that are involved in the interaction with a new multispecific biological drug. This is possible because the large size and the intrinsic flexibility of the complexes are not limiting factors for solid-state NMR.


Asunto(s)
Productos Biológicos , Anticuerpos , Mapeo Epitopo , Espectroscopía de Resonancia Magnética , Proteínas/química
15.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34638887

RESUMEN

Three novel platinum(II) complexes bearing N-heterocyclic ligands, i.e., Pt2c, Pt-IV and Pt-VIII, were previously prepared and characterized. They manifested promising in vitro anticancer properties associated with non-conventional modes of action. To gain further mechanistic insight, we have explored here the reactions of these Pt compounds with a few model proteins, i.e., hen egg white lysozyme (HEWL), bovine pancreatic ribonuclease (RNase A), horse heart cytochrome c (Cyt-c) and human serum albumin (HSA), primarily through ESI MS analysis. Characteristic and variegate patterns of reactivity were highlighted in the various cases that appear to depend both on the nature of the Pt complex and of the interacting protein. The protein-bound Pt fragments were identified. In the case of the complex Pt2c, the adducts formed upon reaction with HEWL and RNase A were further characterized by solving the respective crystal structures: this allowed us to determine the exact location of the various Pt binding sites. The implications of the obtained results are discussed in relation to the possible mechanisms of action of these innovative anticancer Pt complexes.


Asunto(s)
Complejos de Coordinación/química , Citocromos c/química , Muramidasa/química , Platino (Metal)/química , Ribonucleasa Pancreática/química , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Bovinos , Complejos de Coordinación/metabolismo , Cristalografía por Rayos X , Citocromos c/metabolismo , Caballos , Humanos , Ligandos , Modelos Moleculares , Muramidasa/metabolismo , Platino (Metal)/metabolismo , Unión Proteica , Dominios Proteicos , Ribonucleasa Pancreática/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos
16.
Front Genet ; 12: 694468, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34178043

RESUMEN

The development of high-throughput high-content technologies and the increased ease in their application in clinical settings has raised the expectation of an important impact of these technologies on diagnosis and personalized therapy. Patient genomic and expression profiles yield lists of genes that are mutated or whose expression is modulated in specific disease conditions. The challenge remains of extracting from these lists functional information that may help to shed light on the mechanisms that are perturbed in the disease, thus setting a rational framework that may help clinical decisions. Network approaches are playing an increasing role in the organization and interpretation of patients' data. Biological networks are generated by connecting genes or gene products according to experimental evidence that demonstrates their interactions. Till recently most approaches have relied on networks based on physical interactions between proteins. Such networks miss an important piece of information as they lack details on the functional consequences of the interactions. Over the past few years, a number of resources have started collecting causal information of the type protein A activates/inactivates protein B, in a structured format. This information may be represented as signed directed graphs where physiological and pathological signaling can be conveniently inspected. In this review we will (i) present and compare these resources and discuss the different scope in comparison with pathway resources; (ii) compare resources that explicitly capture causality in terms of data content and proteome coverage (iii) review how causal-graphs can be used to extract disease-specific Boolean networks.

17.
Genes (Basel) ; 12(3)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33809949

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has caused more than 2.3 million casualties worldwide and the lack of effective treatments is a major health concern. The development of targeted drugs is held back due to a limited understanding of the molecular mechanisms underlying the perturbation of cell physiology observed after viral infection. Recently, several approaches, aimed at identifying cellular proteins that may contribute to COVID-19 pathology, have been reported. Albeit valuable, this information offers limited mechanistic insight as these efforts have produced long lists of cellular proteins, the majority of which are not annotated to any cellular pathway. We have embarked in a project aimed at bridging this mechanistic gap by developing a new bioinformatic approach to estimate the functional distance between a subset of proteins and a list of pathways. A comprehensive literature search allowed us to annotate, in the SIGNOR 2.0 resource, causal information underlying the main molecular mechanisms through which severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and related coronaviruses affect the host-cell physiology. Next, we developed a new strategy that enabled us to link SARS-CoV-2 interacting proteins to cellular phenotypes via paths of causal relationships. Remarkably, the extensive information about inhibitors of signaling proteins annotated in SIGNOR 2.0 makes it possible to formulate new potential therapeutic strategies. The proposed approach, which is generally applicable, generated a literature-based causal network that can be used as a framework to formulate informed mechanistic hypotheses on COVID-19 etiology and pathology.


Asunto(s)
Autofagia/genética , COVID-19/metabolismo , COVID-19/virología , Interacciones Microbiota-Huesped/genética , SARS-CoV-2/metabolismo , Transducción de Señal , COVID-19/genética , COVID-19/patología , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/virología , Proteoma , PubMed , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Transducción de Señal/genética
18.
Proteomes ; 9(2)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925552

RESUMEN

FLT3 mutations are the most frequently identified genetic alterations in acute myeloid leukemia (AML) and are associated with poor clinical outcome, relapse and chemotherapeutic resistance. Elucidating the molecular mechanisms underlying FLT3-dependent pathogenesis and drug resistance is a crucial goal of biomedical research. Given the complexity and intricacy of protein signaling networks, deciphering the molecular basis of FLT3-driven drug resistance requires a systems approach. Here we discuss how the recent advances in mass spectrometry (MS)-based (phospho) proteomics and multiparametric analysis accompanied by emerging computational approaches offer a platform to obtain and systematically analyze cell-specific signaling networks and to identify new potential therapeutic targets.

19.
J Clin Med ; 10(4)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671425

RESUMEN

The embryonal rhabdomyosarcoma (eRMS) is a soft tissue sarcoma commonly affecting the head and neck, the extremities and the genitourinary tract. To contribute to revealing the cell types that may originate this tumor, we exploited mass cytometry, a single-cell technique that, by using heavy-metal-tagged antibodies, allows the accurate monitoring of the changes occurring in the mononuclear cell composition of skeletal muscle tissue during tumor development. To this end, we compared cell populations of healthy muscles with those from spatiotemporal-induced eRMS tumors in a mouse model (LSL-KrasG12D/+;Tp53Fl/Fl) that can be used to develop rhabdomyosarcoma by means of infection with an adenovirus vector expressing Cre (Ad-Cre) recombinase. By monitoring different time points after tumor induction, we were able to analyze tumor progression and composition, identifying fibro/adipogenic progenitors (FAPs) as the cell type that, in this model system, had a pivotal role in tumor development. In vitro studies highlighted that both FAPs and satellite cells (SCs), upon infection with the Ad-Cre, acquired the potential to develop rhabdomyosarcomas when transplanted into immunocompromised mice. However, only infected FAPs had an antigen profile that was similar to embryonal rhabdomyosarcoma cells. Overall, our analysis supports the involvement of FAPs in eRMS development.

20.
J Pers Med ; 11(2)2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33578936

RESUMEN

High throughput technologies such as deep sequencing and proteomics are increasingly becoming mainstream in clinical practice and support diagnosis and patient stratification. Developing computational models that recapitulate cell physiology and its perturbations in disease is a required step to help with the interpretation of results of high content experiments and to devise personalized treatments. As complete cell-models are difficult to achieve, given limited experimental information and insurmountable computational problems, approximate approaches should be considered. We present here a general approach to modeling complex diseases by embedding patient-specific genomics data into actionable logic models that take into account prior knowledge. We apply the strategy to acute myeloid leukemia (AML) and assemble a network of logical relationships linking most of the genes that are found frequently mutated in AML patients. We derive Boolean models from this network and we show that by priming the model with genomic data we can infer relevant patient-specific clinical features. Here we propose that the integration of literature-derived causal networks with patient-specific data should be explored to help bedside decisions.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...